The Chemosensory Function of Primary Cilia Regulates Cholangiocyte Migration, Invasion, and Tumor Growth

Hepatology. 2019 Apr;69(4):1582-1598. doi: 10.1002/hep.30308. Epub 2019 Mar 15.

Abstract

Cholangiocytes, the epithelial cells lining the biliary tree in the liver, express primary cilia that can detect several kinds of environmental signals and then transmit this information into the cell. We have reported that cilia are significantly reduced in cholangiocarcinoma (CCA) and that the experimental deciliation of normal cells induces a malignant-like phenotype with increased proliferation, anchorage-independent growth, invasion, and migration. Here, we tested the hypothesis that the chemosensory function of cholangiocyte primary cilia acts as a mechanism for tumor suppression. We found that in the presence of extracellular nucleotides cilia-dependent chemosensation of the nucleotides inhibited migration and invasion in normal ciliated cholangiocytes through a P2Y11 receptor and liver kinase B1 (LKB1)-phosphatase and tensin homolog-AKT-dependent mechanism. In contrast, in normal deciliated cholangiocytes and CCA cells, the nucleotides induced the opposite effects, i.e., increased migration and invasion. As activation of LKB1 through a cilia-dependent mechanism was required for the nucleotide-mediated inhibitory effects on migration and invasion, we attempted to activate LKB1 directly, independent of ciliary expression, using the compound hesperidin methyl chalcone (HMC). We found that HMC induced activation of LKB1 in both ciliated and deciliated cells in vitro, resulting in the inhibition of migration and proliferation. Furthermore, using a rat syngeneic orthotopic CCA model, we found that HMC inhibited tumor growth in vivo. Conclusion: These findings highlight the importance of the chemosensory function of primary cilia for the control of migration and invasion and suggest that, by directly activating LKB1 and bypassing the need for primary cilia, it is possible to emulate this chemosensory function in CCA cells; these data warrant further studies evaluating the possibility of using HMC as therapy for CCA.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • AMP-Activated Protein Kinase Kinases
  • Adenosine Triphosphate / metabolism
  • Adenylyl Cyclases / metabolism
  • Animals
  • Bile Duct Neoplasms / etiology*
  • Cell Line, Tumor
  • Cell Movement
  • Chemoreceptor Cells / physiology*
  • Cholangiocarcinoma / etiology*
  • Cilia / physiology*
  • Cyclic AMP-Dependent Protein Kinases / metabolism
  • Humans
  • PTEN Phosphohydrolase / metabolism
  • Protein Serine-Threonine Kinases / metabolism
  • Proto-Oncogene Proteins c-akt / metabolism
  • Rats
  • Receptors, Purinergic P2 / metabolism

Substances

  • P2RY11 protein, human
  • Receptors, Purinergic P2
  • Adenosine Triphosphate
  • Protein Serine-Threonine Kinases
  • Proto-Oncogene Proteins c-akt
  • STK11 protein, human
  • Cyclic AMP-Dependent Protein Kinases
  • AMP-Activated Protein Kinase Kinases
  • PTEN Phosphohydrolase
  • Adenylyl Cyclases
  • adenylyl cyclase type V